Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Cell Microbiol ; 21(6): e13017, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30761726

RESUMO

α-Haemolysin (HlyA) from uropathogenic Escherichia coli has been demonstrated to be a significant virulence factor for ascending urinary tract infections. Once the E. coli reach the well-vascularised kidneys, there is a high risk of bacteraemia and a subsequent septic host response. Despite this, HlyA has the potential to accelerate the host response both directly and via its ability to facilitate adenosine triphosphate release from cells. It has not been settled whether HlyA aggravates bacteraemia into a septic state. To address this, we used an E. coli strain in a model of acute urosepsis that was either transfected with a plasmid containing the full HlyA operon or one with deletion in the HlyA gene. Here, we show that HlyA accelerates the host response to E. coli in the circulation. Mice exposed to HlyA-producing E. coli showed massively increased proinflammatory cytokines, a substantial fall in circulating thrombocytes, extensive haematuria, and intravascular haemolysis. This was not seen in mice exposed to either E. coli that do not secrete HlyA or vehicle controls. Consistent with the massive host response to the bacteria, the mice exposed to HlyA-producing E. coli died exceedingly early, whereas mice exposed to E. coli without HlyA production and vehicle controls survived the entire observation period. These data allow us to conclude that HlyA is a virulence factor that accelerates a state of bacteraemia into fulminant sepsis in a mouse model.


Assuntos
Bacteriemia/microbiologia , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/sangue , Proteínas Hemolisinas/sangue , Infecções Urinárias/microbiologia , Escherichia coli Uropatogênica/patogenicidade , Fatores de Virulência/sangue , Animais , Bacteriemia/sangue , Bacteriemia/mortalidade , Plaquetas/metabolismo , Citocinas/sangue , Modelos Animais de Doenças , Eritrócitos/metabolismo , Eritrócitos/microbiologia , Eritrócitos/patologia , Infecções por Escherichia coli/sangue , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Expressão Gênica , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Hemólise , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Óperon , Infecções Urinárias/sangue , Escherichia coli Uropatogênica/metabolismo , Fatores de Virulência/genética
2.
Int J Mol Sci ; 21(16)2020 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-32781764

RESUMO

Urosepsis is a potentially life-threatening, systemic reaction to uropathogenic bacteria entering the bloodstream of the host. One of the hallmarks of sepsis is early thrombocyte activation with a following fall in circulating thrombocytes as a result of intravascular aggregation and sequestering of thrombocytes in the major organs. Development of a thrombocytopenic state is associated with a poorer outcome of sepsis. Uropathogenic Escherichia coli frequently produce the pore-forming, virulence factor α-haemolysin (HlyA), of which the biological effects are mediated by ATP release and subsequent activation of P2 receptors. Thus, we speculated that inhibition of thrombocyte P2Y1 and P2Y12 receptors might ameliorate the septic response to HlyA-producing E. coli. The study combined in vitro measurements of toxin-induced thrombocyte activation assessed as increased membrane abundance of P-selectin, fibronectin and CD63 and data from in vivo murine model of sepsis-induced by HlyA-producing E. coli under infusion of P2Y1 and P2Y12 antagonists. Our data show that the P2Y1 receptor antagonist almost abolishes thrombocyte activation by pore-forming bacterial toxins. Inhibition of P2Y1, by constant infusion of MRS2500, markedly increased the survival in mice with induced sepsis. Moreover, MRS2500 partially prevented the sepsis-induced depletion of circulating thrombocytes and dampened the sepsis-associated increase in proinflammatory cytokines. In contrast, P2Y12 receptor inhibition had only a marginal effect in vivo and in vitro. Taken together, inhibition of the P2Y1 receptor gives a subtle dampening of the thrombocyte activation and the cytokine response to bacteraemia, which may explain the improved survival observed by P2Y1 receptor antagonists.


Assuntos
Toxinas Bacterianas/toxicidade , Plaquetas/patologia , Receptores Purinérgicos P2Y12/metabolismo , Receptores Purinérgicos P2Y1/metabolismo , Sepse/patologia , Infecções Urinárias/patologia , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/farmacologia , Monofosfato de Adenosina/uso terapêutico , Animais , Nucleotídeos de Desoxiadenina/farmacologia , Modelos Animais de Doenças , Proteínas de Escherichia coli/metabolismo , Proteínas Hemolisinas/metabolismo , Humanos , Masculino , Camundongos Endogâmicos BALB C , Sepse/complicações , Sepse/tratamento farmacológico , Resultado do Tratamento , Infecções Urinárias/complicações , Infecções Urinárias/tratamento farmacológico , Escherichia coli Uropatogênica/efeitos dos fármacos
3.
Purinergic Signal ; 15(2): 265-276, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31129780

RESUMO

Urosepsis is a severe condition often caused by Escherichia coli that spontaneously have ascended the urinary tract to the kidneys causing pyelonephritis and potentially bacteraemia. The number of sepsis cases has been steadily increasing over the last decades, and there are still no specific, molecular supportive therapies for sepsis to supplement antibiotic treatment. P2X1 receptors are expressed by a number of immune cells including thrombocytes, which presently have been established as an important player in the acute immune response to bacterial infections. P2X1 receptor-deficient mice have been shown to be relatively protected against urosepsis, with markedly reduced levels of circulating proinflammatory cytokines and intravascular coagulation. However, here we show that continuous intravenous infusion with P2X1 receptor antagonist markedly accelerates development of a septic response to induced bacteraemia with uropathogenic E. coli. Mice exposed to the P2X1 receptor antagonists die very early with haematuria, substantially elevated plasma levels of proinflammatory cytokines, massive intravascular coagulation and a concomitant reduction in circulating thrombocytes. Interestingly, infusion of P2X1 receptor antagonists causes a marked acute reduction in circulating thrombocytes and a higher number of bacteria in the blood. These data support the notion that the number of functional thrombocytes is important for the acute defence against bacteria in the circulation and that the P2X1 receptor potentially could be essential for this response.


Assuntos
Plaquetas/efeitos dos fármacos , Infecções por Escherichia coli , Antagonistas do Receptor Purinérgico P2X/farmacologia , Receptores Purinérgicos P2X1 , Sepse , Infecções Urinárias , Animais , Benzenossulfonatos , Proteínas Hemolisinas , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pielonefrite , Suramina/análogos & derivados , Escherichia coli Uropatogênica
4.
J Membr Biol ; 250(3): 301-313, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28488084

RESUMO

Uropathogenic Escherichia coli often produce the virulence factor α-hemolysin (HlyA), and the more severe the infection, the likelier it is to isolate HlyA-producing E. coli from patients. HlyA forms pores upon receptor-independent insertion of the toxin into biological membranes and it has been substantiated that HlyA-induced hemolysis is amplified by toxin-induced ATP release and activation of P2X receptors. Thus, hemolysis inflicted by HlyA is a protracted process involving signal transduction. It consists of early, marked cell shrinkage followed by swelling and eventually lysis. The initially shrinkage is a consequence of a substantial Ca2+-influx and activation of Ca2+-sensitive K+ and Cl- channels (KCa3.1/TMEM16A). The shrinkage is followed by gradual cell swelling, which ultimately lyses the cells. These findings clearly show that the HlyA pore provides a substantial volume challenge for the cells, and the fate of the given cell is co-determined by intrinsic erythrocytal volume regulation. We therefore speculated that other mechanisms involved in erythrocyte volume regulation may influence the hemolytic process inflicted by HlyA. Strikingly, HlyA-induced hemolysis is markedly reduced in erythrocytes isolated from NKCC1-deficient (NKCC1-/-) mice compared to controls. The NKCC1 inhibitors furosemide and bumetanide concentration-dependently inhibit HlyA-induced lysis of human and murine erythrocytes. However, in high concentrations bumetanide further reduced hemolysis in erythrocytes from NKCC1-/- mice and, thus, also exhibit indirect effects on hemolysis. The effect of loop diuretics on the hemolysis is not unique to HlyA but is similarly seen in LtxA- and α-toxin-induced hemolysis. Bumetanide clearly potentiates HlyA-induced volume reduction and delays the following erythrocyte swelling. This allows increased phagocytosis of damaged erythrocytes by THP-1 cell as a result of prolonged cell shrinkage. These data suggest that erythrocyte susceptibility to cytolysins is modified by NKCC1 and signifies intrinsic volume regulators as important determinants of cellular outcome of pore-forming toxins.


Assuntos
Escherichia coli/química , Proteínas Hemolisinas/farmacologia , Animais , Proteínas de Bactérias/farmacologia , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Furosemida/farmacologia , Hemólise/efeitos dos fármacos , Humanos , Camundongos , Camundongos Knockout , Fagocitose/efeitos dos fármacos , Membro 2 da Família 12 de Carreador de Soluto/deficiência , Membro 2 da Família 12 de Carreador de Soluto/genética , Membro 2 da Família 12 de Carreador de Soluto/metabolismo , Células THP-1
5.
J Biol Chem ; 290(23): 14776-84, 2015 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-25911098

RESUMO

Urinary tract infections are commonly caused by α-hemolysin (HlyA)-producing Escherichia coli. In erythrocytes, the cytotoxic effect of HlyA is strongly amplified by P2X receptors, which are activated by extracellular ATP released from the cytosol of the erythrocytes. In renal epithelia, HlyA causes reversible [Ca(2+)]i oscillations, which trigger interleukin-6 (IL-6) and IL-8 release. We speculate that this effect is caused by HlyA-induced ATP release from the epithelial cells and successive P2 receptor activation. Here, we demonstrate that HlyA-induced [Ca(2+)]i oscillations in renal epithelia were completely prevented by scavenging extracellular ATP. In accordance, HlyA was unable to inflict any [Ca(2+)]i oscillations in 132-1N1 cells, which lack P2R completely. After transfecting these cells with the hP2Y2 receptor, HlyA readily triggered [Ca(2+)]i oscillations, which were abolished by P2 receptor antagonists. Moreover, HlyA-induced [Ca(2+)]i oscillations were markedly reduced in medullary thick ascending limbs isolated from P2Y2 receptor-deficient mice compared with wild type. Interestingly, the following HlyA-induced IL-6 release was absent in P2Y2 receptor-deficient mice. This suggests that HlyA induces ATP release from renal epithelia, which via P2Y2 receptors is the main mediator of HlyA-induced [Ca(2+)]i oscillations and IL-6 release. This supports the notion that ATP signaling occurs early during bacterial infection and is a key player in the further inflammatory response.


Assuntos
Infecções por Escherichia coli/imunologia , Proteínas de Escherichia coli/imunologia , Escherichia coli/fisiologia , Proteínas Hemolisinas/imunologia , Interleucina-6/imunologia , Rim/microbiologia , Receptores Purinérgicos P2Y2/imunologia , Urotélio/microbiologia , Trifosfato de Adenosina/imunologia , Animais , Sinalização do Cálcio , Linhagem Celular , Cães , Escherichia coli/imunologia , Humanos , Rim/imunologia , Camundongos , Urotélio/imunologia
6.
Infect Immun ; 84(11): 3114-3130, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27528275

RESUMO

α-Hemolysin (HlyA) from Escherichia coli and leukotoxin A (LtxA) from Aggregatibacter actinomycetemcomitans are important virulence factors in ascending urinary tract infections and aggressive periodontitis, respectively. The extracellular signaling molecule ATP is released immediately after insertion of the toxins into plasma membranes and, via P2X receptors, is essential for the erythrocyte damage inflicted by these toxins. Moreover, ATP signaling is required for the ensuing recognition and phagocytosis of damaged erythrocytes by the monocytic cell line THP-1. Here, we investigate how these toxins affect THP-1 monocyte function. We demonstrate that both toxins trigger early ATP release and a following increase in the intracellular Ca2+ concentration ([Ca2+]i) in THP-1 monocytes. The HlyA- and LtxA-induced [Ca2+]i response is diminished by the P2 receptor antagonist in a pattern that fits the functional P2 receptor expression in these cells. Both toxins are capable of lysing THP-1 cells, with LtxA being more aggressive. Either desensitization or blockage of P2X1, P2X4, or P2X7 receptors markedly reduces toxin-induced cytolysis. This pattern is paralleled in freshly isolated human monocytes from healthy volunteers. Interestingly, only a minor fraction of the toxin-damaged THP-1 monocytes eventually lyse. P2X7 receptor inhibition generally prevents cell damage, except from a distinct cell shrinkage that prevails in response to the toxins. Moreover, we find that preexposure to HlyA preserves the capacity of THP-1 monocytes to phagocytose damaged erythrocytes and may induce readiness to discriminate between damaged and healthy erythrocytes. These findings suggest a new pharmacological target for protecting monocytes during exposure to pore-forming cytolysins during infection or injury.


Assuntos
Aggregatibacter actinomycetemcomitans/metabolismo , Toxinas Bacterianas/toxicidade , Escherichia coli/metabolismo , Proteínas Hemolisinas/toxicidade , Monócitos/efeitos dos fármacos , Receptores Purinérgicos P2X/fisiologia , Toxinas Bacterianas/metabolismo , Morte Celular/efeitos dos fármacos , Citoplasma/metabolismo , Citotoxinas/metabolismo , Eritrócitos/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/toxicidade , Exotoxinas/metabolismo , Exotoxinas/toxicidade , Proteínas Hemolisinas/metabolismo , Hemólise/fisiologia , Humanos , Monócitos/metabolismo
7.
J Biol Chem ; 289(27): 19098-109, 2014 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-24860098

RESUMO

ATP is as an extracellular signaling molecule able to amplify the cell lysis inflicted by certain bacterial toxins including the two RTX toxins α-hemolysin (HlyA) from Escherichia coli and leukotoxin A (LtxA) from Aggregatibacter actinomycetemcomitans. Inhibition of P2X receptors completely blocks the RTX toxin-induced hemolysis over a larger concentration range. It is, however, at present not known how the ATP that provides the amplification is released from the attacked cells. Here we show that both HlyA and LtxA trigger acute release of ATP from human erythrocytes that preceded and were not caused by cell lysis. This early ATP release did not occur via previously described ATP-release pathways in the erythrocyte. Both HlyA and LtxA were capable of triggering ATP release in the presence of the pannexin 1 blockers carbenoxolone and probenecid, and the HlyA-induced ATP release was found to be similar in erythrocytes from pannexin 1 wild type and knock-out mice. Moreover, the voltage-dependent anion channel antagonist TRO19622 had no effect on ATP release by either of the toxins. Finally, we showed that both HlyA and LtxA were able to release ATP from ATP-loaded lipid (1-palmitoyl-2-oleoyl-phosphatidylcholine) vesicles devoid of any erythrocyte channels or transporters. Again we were able to show that this happened in a non-lytic fashion, using calcein-containing vesicles as controls. These data show that both toxins incorporate into lipid vesicles and allow ATP to be released. We suggest that both toxins cause acute ATP release by letting ATP pass the toxin pores in both human erythrocytes and artificial membranes.


Assuntos
Trifosfato de Adenosina/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/farmacologia , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/farmacologia , Proteínas Hemolisinas/química , Proteínas Hemolisinas/farmacologia , Aggregatibacter actinomycetemcomitans , Animais , Conexinas/deficiência , Conexinas/genética , Eritrócitos/citologia , Técnicas de Inativação de Genes , Hemoglobinas/metabolismo , Hemólise/efeitos dos fármacos , Humanos , Membranas Artificiais , Camundongos , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Fosfatidilcolinas/metabolismo , Porosidade
8.
Infect Immun ; 82(6): 2219-28, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24643533

RESUMO

Leukotoxin (LtxA) from Aggregatibacter actinomycetemcomitans is known to target and lyse ß2-integrin-expressing cells such as polymorphonuclear leukocytes and macrophages. LtxA is an important virulence factor that facilitates chronic inflammation and is strongly associated with a fast-progressing form of periodontitis caused by the JP2 clone of the bacterium. Here, we show that sialic acid residues are important for LtxA-induced cell lysis, regardless of whether the cell express ß2-integrin or not. Clearly, removal of sialic acid groups significantly reduces a ß2-integrin-specific LtxA-induced lysis. Moreover, sialic acid presented on alternative proteins, such as, for instance, on erythrocytes that do not express ß2-integrin, also makes the cells more sensitive to LtxA. The data also illustrate the importance of the negative charge in order for the sialic acid to associate LtxA with the membrane. Removal of sialic acid is in itself sufficient to significantly reduce the negative charge on the erythrocytes. Moreover, we found that on human erythrocytes there is a positive association between the sensitivity to LtxA and the amount of negative charge caused by sialic acid. Interestingly, these features are not shared by all RTX toxins, since α-hemolysin from Escherichia coli induced cell lysis of both ß2-integrin-expressing and nonexpressing cells and this lysis is independent of the presence of sialic acid residues. In conclusion, LtxA not only is cytotoxic to ß2-integrin-expressing cells but can potentially initiate cell lysis in all cells that present a sufficient density of sialic acid groups on their plasma membrane.


Assuntos
Aggregatibacter actinomycetemcomitans/fisiologia , Morte Celular/fisiologia , Eritrócitos/efeitos dos fármacos , Exotoxinas/fisiologia , Ácido N-Acetilneuramínico/fisiologia , Análise de Variância , Animais , Antígenos CD18/fisiologia , Morte Celular/efeitos dos fármacos , Linhagem Celular , Eritrócitos/metabolismo , Exotoxinas/toxicidade , Humanos , Camundongos , Ácido N-Acetilneuramínico/química , Coelhos , Ovinos
9.
Sci Transl Med ; 16(739): eadk9109, 2024 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-38507469

RESUMO

Myasthenia gravis (MG) is a neuromuscular disease that results in compromised transmission of electrical signals at the neuromuscular junction (NMJ) from motor neurons to skeletal muscle fibers. As a result, patients with MG have reduced skeletal muscle function and present with symptoms of severe muscle weakness and fatigue. ClC-1 is a skeletal muscle specific chloride (Cl-) ion channel that plays important roles in regulating neuromuscular transmission and muscle fiber excitability during intense exercise. Here, we show that partial inhibition of ClC-1 with an orally bioavailable small molecule (NMD670) can restore muscle function in rat models of MG and in patients with MG. In severely affected MG rats, ClC-1 inhibition enhanced neuromuscular transmission, restored muscle function, and improved mobility after both single and prolonged administrations of NMD670. On this basis, NMD670 was progressed through nonclinical safety pharmacology and toxicology studies, leading to approval for testing in clinical studies. After successfully completing phase 1 single ascending dose in healthy volunteers, NMD670 was tested in patients with MG in a randomized, placebo-controlled, single-dose, three-way crossover clinical trial. The clinical trial evaluated safety, pharmacokinetics, and pharmacodynamics of NMD670 in 12 patients with mild MG. NMD670 had a favorable safety profile and led to clinically relevant improvements in the quantitative myasthenia gravis (QMG) total score. This translational study spanning from single muscle fiber recordings to patients provides proof of mechanism for ClC-1 inhibition as a potential therapeutic approach in MG and supports further development of NMD670.


Assuntos
Cloretos , Miastenia Gravis , Humanos , Ratos , Animais , Cloretos/uso terapêutico , Miastenia Gravis/tratamento farmacológico , Músculo Esquelético/fisiologia , Junção Neuromuscular , Canais de Cloreto
10.
Cell Microbiol ; 14(12): 1904-20, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22906303

RESUMO

Leukotoxin (LtxA) is a virulence factor secreted by the bacterium Aggregatibacter actinomycetemcomitans, which can cause localized aggressive periodontitis and endocarditis. LtxA belongs to the repeat-in-toxin (RTX) family of exotoxins of which other members inflict lysis by formation of membrane pores. Recently, we documented that the haemolytic process induced by another RTX toxin [α-haemolysin (HlyA) from Escherichia coli] requires P2X receptor activation and consists of sequential cell shrinkage and swelling. In contrast, the cellular and molecular mechanisms of LtxA-mediated haemolysis are not fully understood. Here, we investigate the effect of LtxA on erythrocyte volume and whether P2 receptors also play a part in LtxA-mediated haemolysis. We observed that LtxA initially decreases the cell size, followed by a gradual rise in volume until the cell finally lyses. Moreover, LtxA triggers phosphatidylserine (PS) exposure in the erythrocyte membrane and both the shrinkage and the PS-exposure is preceded by increments in the intracellular Ca(2+) concentration ([Ca(2+)](i)). Interestingly, LtxA-mediated haemolysis is significantly potentiated by ATP release and P2X receptor activation in human erythrocytes. Furthermore, the LtxA-induced [Ca(2+)](i) increase and following volume changes partially depend on P2 receptor activation. Theseobservations imply that intervention against local P2-mediated auto- and paracrine signalling may prevent LtxA-mediated cell damage.


Assuntos
Eritrócitos/efeitos dos fármacos , Exotoxinas/toxicidade , Hemólise , Pasteurellaceae/patogenicidade , Receptores Purinérgicos P2X/metabolismo , Cálcio/análise , Tamanho Celular , Citoplasma/química , Eritrócitos/citologia , Escherichia coli , Humanos , Modelos Biológicos
11.
Proc Natl Acad Sci U S A ; 106(10): 4030-5, 2009 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-19225107

RESUMO

Escherichia coli is the dominant facultative bacterium in the normal intestinal flora. E. coli is, however, also responsible for the majority of serious extraintestinal infections. There are distinct serotypical differences between facultative and invasive E. coli strains. Invasive strains frequently produce virulence factors such as alpha-hemolysin (HlyA), which causes hemolysis by forming pores in the erythrocyte membrane. The present study reveals that this pore formation triggers purinergic receptor activation to mediate the full hemolytic action. Non-selective ATP-receptor (P2) antagonists (PPADS, suramin) and ATP scavengers (apyrase, hexokinase) concentration dependently inhibited HlyA-induced lysis of equine, murine, and human erythrocytes. The pattern of responsiveness to more selective P2-antagonists implies that both P2X(1) and P2X(7) receptors are involved in HlyA-induced hemolysis in all three species. In addition, our results also propose a role for the pore protein pannexin1 in HlyA-induced hemolysis, as non-selective inhibitors of this channel significantly reduced hemolysis in the three species. In conclusion, activation of P2X receptors and possibly also pannexins augment hemolysis induced by the bacterial toxin, HlyA. These findings potentially have clinical perspectives as P2 antagonists may ameliorate symptoms during sepsis with hemolytic bacteria.


Assuntos
Proteínas de Escherichia coli/farmacologia , Escherichia coli/crescimento & desenvolvimento , Proteínas Hemolisinas/farmacologia , Hemólise/efeitos dos fármacos , Receptores Purinérgicos P2/metabolismo , Adenosina Trifosfatases/farmacologia , Animais , Conexinas/antagonistas & inibidores , Eritrócitos/citologia , Eritrócitos/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Cavalos , Humanos , Camundongos , Antagonistas do Receptor Purinérgico P2 , Receptores Purinérgicos P2X
12.
J Biol Chem ; 285(20): 15557-15565, 2010 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-20231275

RESUMO

alpha-Hemolysin from Escherichia coli (HlyA) readily lyse erythrocytes from various species. We have recently demonstrated that this pore-forming toxin provokes distinct shrinkage and crenation before it finally leads to swelling and lysis of erythrocytes. The present study documents the underlying mechanism for this severe volume reduction. We show that HlyA-induced shrinkage and crenation of human erythrocytes occur subsequent to a significant rise in [Ca(2+)](i). The Ca(2+)-activated K(+) channel K(Ca)3.1 (or Gardos channel) is essential for the initial shrinkage, because both clotrimazole and TRAM-34 prevent the shrinkage and potentiate hemolysis produced by HlyA. Notably, the recently described Ca(2+)-activated Cl(-) channel TMEM16A contributes substantially to HlyA-induced cell volume reduction. Erythrocytes isolated from TMEM16A(-/-) mice showed significantly attenuated crenation and increased lysis compared with controls. Additionally, we found that HlyA leads to acute exposure of phosphatidylserine in the outer leaflet of the plasma membrane. This exposure was considerably reduced by K(Ca)3.1 antagonists. In conclusion, this study shows that HlyA triggers acute erythrocyte shrinkage, which depends on Ca(2+)-activated efflux of K(+) via K(Ca)3.1 and Cl(-) via TMEM16A, with subsequent phosphatidylserine exposure. This mechanism might potentially allow HlyA-damaged erythrocytes to be removed from the bloodstream by macrophages and thereby reduce the risk of intravascular hemolysis.


Assuntos
Tamanho Celular , Eritrócitos/efeitos dos fármacos , Proteínas de Escherichia coli/farmacologia , Proteínas Hemolisinas/farmacologia , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Neoplasias/efeitos dos fármacos , Fosfatidilserinas/farmacologia , Canais de Potássio Cálcio-Ativados/efeitos dos fármacos , Animais , Anoctamina-1 , Células Cultivadas , Canais de Cloreto , Citometria de Fluxo , Hemólise/efeitos dos fármacos , Humanos , Proteínas de Membrana/fisiologia , Camundongos , Proteínas de Neoplasias/fisiologia , Canais de Potássio Cálcio-Ativados/fisiologia
13.
Pflugers Arch ; 462(5): 669-79, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21847558

RESUMO

Recently, it was documented that α-haemolysin (HlyA) from Escherichia coli uses erythrocyte P2 receptors cause lysis. This finding was surprising as it appeared firmly established that HlyA-dependent pore formation per se is sufficient for full cell lysis. We discovered that HlyA induced a sequential process of shrinkage and swelling and that the final haemolysis is completely prevented by blockers of P2X receptors and pannexin channels. This finding has potential clinical relevance as it may offer specific pharmacological interference to ameliorate haemolysis inflicted by pore-forming bacterial toxins. In this context, it is essential to know whether this is specific to HlyA-induced cell damage or if other bacterial pore-forming toxins involve purinergic signals to orchestrate haemolysis. Here, we investigate if the haemolysis produced by α-toxin from Staphylococcus aureus involves P2 receptor activation. We observed that α-toxin-induced haemolysis is completely blocked by the unselective P2 receptor antagonist pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid. Moreover, several selective blockers of P2X(1) and P2X(7) ionotropic receptors abolished haemolysis in murine and equine erythrocytes. Inhibitors of pannexin channels partially reduced the α-toxin induced lysis. Thus, we conclude that α-toxin, similar to HlyA from E. coli produces cell damage by specific activation of a purinergic signalling cascade. These data indicate that pore-forming toxins in general require purinergic signalling to elicit their toxicity.


Assuntos
Toxinas Bacterianas/toxicidade , Proteínas Hemolisinas/toxicidade , Hemólise/efeitos dos fármacos , Receptores Purinérgicos P2X/fisiologia , Animais , Carbenoxolona/farmacologia , Conexinas/antagonistas & inibidores , Proteínas de Escherichia coli/toxicidade , Feminino , Cavalos , Humanos , Masculino , Camundongos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Fosfato de Piridoxal/análogos & derivados , Fosfato de Piridoxal/farmacologia , Receptores Purinérgicos P2X/efeitos dos fármacos , Receptores Purinérgicos P2X7/fisiologia
14.
J Membr Biol ; 244(3): 131-40, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22064954

RESUMO

α-Hemolysin (HlyA) from Escherichia coli lyses mammalian erythrocytes by creating nonselective cation pores in the membrane. Pore insertion triggers ATP release and subsequent P2X receptor and pannexin channel activation. Blockage of either P2X receptors or pannexin channels reduces HlyA-induced hemolysis. We found that erythrocytes from Python regius and Python molurus are remarkably resistant to HlyA-induced hemolysis compared to human and Trachemys scripta erythrocytes. HlyA concentrations that induced maximal hemolysis of human erythrocytes did not affect python erythrocytes, but increasing the HlyA concentration 40-fold did induce hemolysis. Python erythrocytes were more resistant to osmotic stress than human erythrocytes, but osmotic stress tolerance per se did not confer HlyA resistance. Erythrocytes from T. scripta, which showed higher osmotic resistance than python erythrocytes, were as susceptible to HlyA as human erythrocytes. Therefore, we tested whether python erythrocytes lack the purinergic signalling known to amplify HlyA-induced hemolysis in human erythrocytes. P. regius erythrocytes increased intracellular Ca²âº concentration and reduced cell volume when exposed to 3 mM ATP, indicating the presence of a P2X7-like receptor. In addition, scavenging extracellular ATP or blocking P2 receptors or pannexin channels reduced the HlyA-induced hemolysis. We tested whether the low HlyA sensitivity resulted from low affinity of HlyA to the python erythrocyte membrane. We found comparable incorporation of HlyA into human and python erythrocyte membranes. Taken together, the remarkable HlyA resistance of python erythrocytes was not explained by increased osmotic resistance, lack of purinergic hemolysis amplification, or differences in HlyA affinity.


Assuntos
Proteínas de Bactérias/farmacologia , Eritrócitos/efeitos dos fármacos , Escherichia coli/química , Proteínas Hemolisinas/farmacologia , Animais , Boidae , Células Cultivadas , Eritrócitos/metabolismo , Hemólise/efeitos dos fármacos , Humanos , Transdução de Sinais/efeitos dos fármacos
15.
Acta Oncol ; 49(7): 922-33, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20831479

RESUMO

UNLABELLED: Positron emission tomography (PET) allows non-invasive detection and mapping of tumor hypoxia. However, slow tracer kinetics and low resolution, results in limited tumor-to-normal tissue contrast and the risk of missing areas where hypoxic cells are intermixed with necrosis. The shape of tumor time activity curves (TACs), as deduced from dynamic scans, may allow further separation of tumors/tumor sub-volumes that are inseparable based on static scans. This study was designed to define the added value of dynamic scans. MATERIAL AND METHODS: Three squamous cell carcinoma tumor models were grown in mice. Mice were injected with the (18)F-labeled PET hypoxia-tracer fluoroazomycin arabinoside (FAZA) and the immunologically-detectable hypoxia-marker pimonidazole, and PET scanned dynamically for three to six hours. Subsequently, microregional tracer retention (autoradiography) and the distribution of pimonidazole-retaining cells (immunohistology) and necrosis were analyzed in tumor tissue sections. Dynamic PET data were analysed based on a two-compartment model with irreversible tracer binding generating estimates of the putative hypoxia surrogate markers k(3) (tracer trapping rate constant) and K(i) (influx rate constant from plasma into irreversible bound tracer). RESULTS/DISCUSSION: High tumor-to-reference tissue ratios and a strong linear correlation (R∼0.7 to 0.95) between density of hypoxic cells and FAZA concentration was observed three hours after tracer administration, suggesting that late time PET images provides an accurate measure of hypoxia against which kinetic model estimates can be validated. Tumor TACs varied widely (ranging from distinctly wash-out to accumulative type) among tumor types although pimonidazole-stainings revealed extensive hypoxia in all models. Kinetic analysis of tumor sub-volumes showed that k(3) correlated poorly with late time FAZA retention regionally in two of the three tumor models. The influx rate constant K(i) displayed far less variability and correlated strongly with late time FAZA retention (hypoxia) in two of three tumor models, whereas a non-consistent relationship was observed in the last tumor model. Our study demonstrates the potential usefulness of dynamic PET, but also that a simple two-compartment model may be inappropriate in some tumor models.


Assuntos
Hipóxia/diagnóstico por imagem , Neoplasias/diagnóstico por imagem , Neoplasias/metabolismo , Nitroimidazóis/farmacocinética , Tomografia por Emissão de Pósitrons/métodos , Animais , Carcinoma de Células Escamosas/complicações , Carcinoma de Células Escamosas/diagnóstico por imagem , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Feminino , Neoplasias Gengivais/complicações , Neoplasias Gengivais/diagnóstico por imagem , Neoplasias Gengivais/metabolismo , Neoplasias Gengivais/patologia , Humanos , Hipóxia/complicações , Hipóxia/metabolismo , Hipóxia/patologia , Camundongos , Camundongos Endogâmicos C3H , Transplante de Neoplasias , Neoplasias/complicações , Transplante Heterólogo , Células Tumorais Cultivadas , Neoplasias do Colo do Útero/complicações , Neoplasias do Colo do Útero/diagnóstico por imagem , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/patologia
16.
Fluids Barriers CNS ; 17(1): 2, 2020 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-31906971

RESUMO

BACKGROUND: Genetic disruption of slc4a10, which encodes the sodium-dependent chloride/bicarbonate exchanger Ncbe, leads to a major decrease in Na+-dependent HCO3- import into choroid plexus epithelial cells in mice and to a marked reduction in brain intraventricular fluid volume. This suggests that Ncbe functionally is a key element in vectorial Na+ transport and thereby for cerebrospinal fluid secretion in the choroid plexus. However, slc4a10 disruption results in severe changes in expression of Na+,K+-ATPase complexes and other major transport proteins, indicating that profound cellular changes accompany the genetic manipulation. METHODS: A tandem mass tag labeling strategy was chosen for quantitative mass spectrometry. Alterations in the broader patterns of protein expression in the choroid plexus in response to genetic disruption of Ncbe was validated by semi-quantitative immunoblotting, immunohistochemistry and morphometry. RESULTS: The abundance of 601 proteins were found significantly altered in the choroid plexus from Ncbe ko mice relative to Ncbe wt. In addition to a variety of transport proteins, particularly large changes in the abundance of proteins involved in cellular energy metabolism were detected in the Ncbe ko mice. In general, the abundance of rate limiting glycolytic enzymes and several mitochondrial enzymes were reduced following slc4a10 disruption. Surprisingly, this was accompanied by increased ATP levels in choroid plexus cells, indicating that the reduction in capacity for energy metabolism was adaptive to high ATP rather than causal for a decreased capacity for ion and water transport. Ncbe-deficient cells also had a reduced cell area and decreased K+ content. CONCLUSION: Our findings suggest that the lack of effective Na+-entry into the epithelial cells of the choroid plexus leads to a profound change in the cellular phenotype, shifting from a high-rate secretory function towards a more dormant state; similar to what is observed during ageing or Alzheimer's disease.


Assuntos
Antiportadores de Cloreto-Bicarbonato/genética , Plexo Corióideo/metabolismo , Epitélio/metabolismo , Transporte de Íons/fisiologia , Simportadores de Sódio-Bicarbonato/genética , Animais , Transporte Biológico/genética , Transporte Biológico/fisiologia , Antiportadores de Cloreto-Bicarbonato/fisiologia , Células Epiteliais/metabolismo , Concentração de Íons de Hidrogênio , Transporte de Íons/genética , Camundongos Knockout
17.
Artigo em Inglês | MEDLINE | ID: mdl-28428949

RESUMO

α-haemolysin (HlyA)-producing Escherichia coli commonly inflict severe urinary tract infections, including pyelonephritis, which comprises substantial risk for sepsis. In vitro, the cytolytic effect of HlyA is mainly mediated by ATP release through the HlyA pore and subsequent P2X1/P2X7 receptor activation. This amplification of the lytic process is not unique to HlyA but is observed by many other pore-forming proteins including complement-induced haemolysis. Since free hemoglobin in the blood is known to be associated with a worse outcome in sepsis one could speculate that inhibition of P2X receptors would ameliorate the course of sepsis. Surprisingly, this study demonstrates that [Formula: see text] and [Formula: see text] mice are exceedingly sensitive to sepsis with uropathogenic E. coli. These mice have markedly lower survival, higher cytokine levels and activated intravascular coagulation. Quite the reverse is seen in [Formula: see text] mice, which had markedly lower cytokine levels and less coagulation activation compared to controls after exposure to uropathogenic E. coli. The high cytokine levels in the [Formula: see text] mouse are unexpected, since P2X7 is implicated in caspase-1-dependent IL-1ß production. Here, we demonstrate that IL-1ß production during sepsis with uropathogenic E. coli is mediated by caspase-8, since caspase-8 and RIPK3 double knock out mice show substantially lower cytokine during sepsis and increased survival after injection of TNFα. These data support that P2X7 and P2X4 receptor activation has a protective effect during severe E. coli infection.


Assuntos
Suscetibilidade a Doenças , Infecções por Escherichia coli/patologia , Receptores Purinérgicos P2X1/deficiência , Receptores Purinérgicos P2X4/deficiência , Receptores Purinérgicos P2X7/deficiência , Sepse/patologia , Animais , Modelos Animais de Doenças , Infecções por Escherichia coli/genética , Camundongos , Camundongos Knockout , Análise de Sobrevida , Resultado do Tratamento
18.
Toxins (Basel) ; 5(3): 472-87, 2013 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-23462688

RESUMO

The pore-forming exotoxin α-hemolysin from E. coli causes a significant volume reduction of human erythrocytes that precedes the ultimate swelling and lysis. This shrinkage results from activation of Ca2+-sensitive K+ (KCa3.1) and Cl- channels (TMEM16A) and reduced functions of either of these channels potentiate the HlyA-induced hemolysis. This means that Ca2+-dependent activation of KCa3.1 and TMEM16A protects the cells against early hemolysis. Simultaneous to the HlyA-induced shrinkage, the erythrocytes show increased exposure of phosphatidylserine (PS) in the outer plasma membrane leaflet, which is known to be a keen trigger for phagocytosis. We hypothesize that exposure to HlyA elicits removal of the damaged erythrocytes by phagocytic cells. Cultured THP-1 cells as a model for erythrocytal phagocytosis was verified by a variety of methods, including live cell imaging. We consistently found the HlyA to very potently trigger phagocytosis of erythrocytes by THP-1 cells. The HlyA-induced phagocytosis was prevented by inhibition of KCa3.1, which is known to reduce PS-exposure in human erythrocytes subjected to both ionomycin and HlyA. Moreover, we show that P2X receptor inhibition, which prevents the cell damages caused by HlyA, also reduced that HlyA-induced PS-exposure and phagocytosis. Based on these results, we propose that erythrocytes, damaged by HlyA-insertion, are effectively cleared from the blood stream. This mechanism will potentially reduce the risk of intravascular hemolysis.


Assuntos
Toxinas Bacterianas/toxicidade , Citofagocitose/efeitos dos fármacos , Eritrócitos/efeitos dos fármacos , Proteínas de Escherichia coli/toxicidade , Proteínas Hemolisinas/toxicidade , Hemólise/efeitos dos fármacos , Receptores Purinérgicos P2X/fisiologia , Técnicas de Cultura de Células , Linhagem Celular , Forma Celular/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Eritrócitos/citologia , Eritrócitos/metabolismo , Citometria de Fluxo , Corantes Fluorescentes , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/antagonistas & inibidores , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Macrófagos/citologia , Bloqueadores dos Canais de Potássio/farmacologia , Antagonistas do Receptor Purinérgico P2X/farmacologia , Receptores Purinérgicos P2X/metabolismo
19.
Vascul Pharmacol ; 54(1-2): 13-7, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21040805

RESUMO

PURPOSE: Combretastatin A-4 phosphate (CA4P) is a promising vascular disrupting agent in cancer treatment, but elicits hypertension in patients. The aim of this study was to use a mouse model to investigate whether hypertension or its modification influenced the treatment efficacy of CA4P in combination with other therapies. MATERIAL AND METHODS: C3H mammary carcinoma bearing or non-bearing CDF1 mice were used. The effects of CA4P alone or in combination with the antihypertensive drug hydralazine (HDZ) on mean arterial blood pressure (MABP), hematocrit (Hct) and hemoglobin concentration ([Hb]) were characterized in non-tumor-bearing animals. Tumor-bearing mice were also treated locally with radiation and/or hyperthermia (41.5 ° C; 60 min) in combination with CA4P alone or CA4P plus HDZ, and TCD50 values (radiation dose that controls 50% of tumors) determined. RESULTS: Hct, [Hb] and MABP respectively increased from 49.3 ± 0.3%, 9.1 ± 0.1mM and 110 ± 7 mm Hg, to 54.7 ± 0.2%, 10.3 ± 0.1 mM and 127 ± 5 mm Hg, within 1h after injecting 100 mg/kg CA4P. For each parameter the magnitude of the peak increase was largely dose independent within the CA4P dose range tested (10-250 mg/kg). However, high CA4P doses delayed the return to baseline and Hct and [Hb] recovered more slowly than MABP. Co-administration of HDZ (0.2mg/kg) was able to suppress the CA4P-induced increase in MABP for several hours but did not noticeably affect the changes in Hct and [Hb]. The TCD50 value (± 95% confidence interval) for radiation alone was 53 (51-55) Gy. Tumor irradiation followed by injection of either CA4P (100 mg/kg) or CA4P+HDZ 30 min later reduced the TCD50 values to 50 (46-54) Gy and 48 (45-52) Gy, respectively. Heating tumors after irradiating further decreased the TCD50 value to 46 (43-48) Gy. When all treatments were combined the TCD50 was 35 (32-38) Gy, regardless of whether the drugs were CA4P or CA4P+HDZ. CONCLUSIONS: CA4P significantly increased Hct, [Hb] and MABP. Hypertension, but not increases in Hct and [Hb], could be reversed with the antihypertensive drug HDZ. CA4P significantly improved tumor response to radiation or thermoradiation, neither of which was influenced by the addition of HDZ.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Hipertensão/induzido quimicamente , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Estilbenos/uso terapêutico , Animais , Anti-Hipertensivos/uso terapêutico , Antineoplásicos Fitogênicos/toxicidade , Pressão Sanguínea , Terapia Combinada , Feminino , Hematócrito , Hemoglobinas/análise , Hidralazina/uso terapêutico , Hipertensão/tratamento farmacológico , Neoplasias Mamárias Experimentais/irrigação sanguínea , Camundongos , Estilbenos/toxicidade
20.
J Exp Biol ; 209(Pt 20): 4167-73, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17023609

RESUMO

Synbranchus marmoratus is a facultative air-breathing fish, which uses its buccal cavity as well as its gills for air-breathing. S. marmoratus shows a very pronounced tachycardia when it surfaces to air-breathe. An elevation of heart rate decreases cardiac filling time and therefore may cause a decline in stroke volume (V(S)), but this can be compensated for by an increase in venous tone to maintain stroke volume. Thus, the study on S. marmoratus was undertaken to investigate how stroke volume and venous function are affected during air-breathing. To this end we measured cardiac output (Q), heart rate (f(H)), central venous blood pressure (P(CV)), mean circulatory filling pressure (MCFP), and dorsal aortic blood pressures (P(DA)) in S. marmoratus. Measurements were performed in aerated water (P(O2)>130 mmHg), when the fish alternated between gill ventilation and prolonged periods of apnoeas, as well as during hypoxia (P(O2)

Assuntos
Smegmamorpha/fisiologia , Ar , Animais , Sistema Cardiovascular/efeitos dos fármacos , Sistema Cardiovascular/fisiopatologia , Epinefrina/farmacologia , Brânquias/fisiologia , Hipóxia/fisiopatologia , Receptores Adrenérgicos alfa/fisiologia , Receptores Adrenérgicos beta/fisiologia , Respiração , Veias/fisiologia , Água
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA